• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/274

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

274 Cards in this Set

  • Front
  • Back
What is the neurotransmitter at found at all autonomic ganglia?
Acetylcholine
Where is the origin of the postganglionic sympathetic nerves found?
Paravertebrally near the CNS, along side the thoracolumbar region of the spine
Where is the origin of the postganglionic parasympathetic nerves found?
In the craniosacral regions close to their target organs
Which branch of the ANS does a cholinomimetic agent work on?
The parasympathetic branch and it works to stimulate through muscarinic receptors.
Which branch of the ANS does a cholinolytic agent work on?
The parasympathetic branch and it works to inhibit through blocking the action of muscarinic receptors.
What are the three roles of acetylcholine in the nervous system?
1) NT at all autonomic ganglia
2) NT of the parasympathetic effector sites where it acts on muscarinic receptors
3) At the neuro-myelo junction at muscles
Explaion where are the main baroreceptors for blood pressure located and how can they become dysfunctional?
They are located in the carotid sinus and the aortic arch. They can become dysfunctional through atherosclerosis and calcification, by hardening and not being able to stretch and sense pressure change like they normally would
What two jobs can the vagus (X) nerve do in the heart?
Slow the firing of the SA node (heart rate) and slow the conduction from the SA to the AV (communication)
Does the parasympathetic system have more control over the ventricles or the atria?
The atria and the rest of the region. There is very little parasympathetic innervation below the AV groove.
Which system has a larger ratio of preganglionic to post ganglionic fibers: parasympathetic or sympathetic?
Parasympathetic 1:1! vs 1:20 for sympathetic
Compare the NTs and receptors found at target tissues in the sympathetic and parasympathetic systems.
The sympathetic system uses Norepinephrine and Adrenergic receptors (alpha and Beta); the parasympathetic system uses Acetylcholine and Cholinergic Muscarinic receptors
Compare the NTs and receptors found in the autonomic ganglia of the sympathetic and parasympathetic systems.
Both systems use Acetylcholine and Cholinergic Nicotinic receptors
What is the exception to the rule that the sympathetic nervous system uses NE and Adrenergic receptors in its target tissues?
The Sweat Glands where the sympathetic nervous system uses Acetylcholine and Cholinergic Muscarinic receptors
What NT/receptor combo is used by the sympathetic nervous system to target the the Renal vascular smooth muscle?
Dopamine and the Dopaminergic receptor
Which NTs does the sympathetic nervous system use when sending messages from the adrenal medulla?
Epinephrine and Norepinephrine
What NT/receptor combo does the somatic nervous system use at its target tissues?
Acetylcholine and Nicotinic receptors
What is the main source of fuel for the heart?
Free fatty acids; this is why the sympathetic nervous system has a lipolytic effect during the fight or flight response
Where in the body do we find muscarinic receptors?
In the target tissues of the parasympathetic system, and in sweat glands as targets of the sympathetic system
Where in the body do we find nicotinic receptors?
In all autonomic ganglia, and in the neuro-muscular junction of the somatic nervous system
Where is acetylcholinesterase and butyrlcholinesterase found in the body?
Acetylcholinesterase is ubiquitous at synaptic junctions and butyrlcholinesterase is found in the blood.
What is Pilocarpine used for?
Glaucoma
What is the ion that controls the stimulation of the SA and AV nodes?
Ca++
How does parasympathetic stimulation affect the resting potential of the SA and AV nodes?
It makes it more negative and less likely to reach threshold, thereby slowing down firing and conduction rates
What effects do the muscarinic receptors of the heart have on SA function and AV function?
They slow rate of firing of the SA node and decrease the conduction of the AV node
What effects do the B1 adrenergic receptors in the heart have on SA and AV function?
They increase the rate of firing of the SA node and increase the conduction of the AV node, as well as increase the overall contractility of the heart
What is the second messenger system of the B1 receptor in the myocyte?
cAMP
Sympathetic tone will constrict most blood vessels through activation of which receptor?
Alpha 1
Sympathetic tone will cause vasodilation in most BVs found in what type of body tissue, due to which receptor?
Skeletal muscle; Beta 2
What type of receptors are curiously found in all vascular beds and why is it curious?
Muscarinic receptors; this is curious because muscarinic receptors are mainly use by the parasympathetic nervous system, but there is not much parasympathetic innervation that occurs in the vascular beds
BV tone is mainly regulated by which receptor of the sympathetic nervous system?
Alpha 1
During the fight or flight response what is responsible for sustained vasodilation in skeletal muscle long after the initial effects of the sympathetic response?
Adenosine from the breakdown of ATP
Which sympathetic receptors are present in bronchial glands and what do they do?
Alpha 1 receptors; decrease secretion
Which parasympathetic receptors are present in bronchial glands and what do they do?
Muscarinic receptors; increase secretion
Which sympathetic receptors are present in bronchial smooth muscle and what do they do?
Beta 2; bronchodilate
Which parasympathetic receptors are present in bronchial smooth muscle and what do they do?
Muscarinic; bronchoconstrict
Which sympathetic receptors are present in GI smooth muscle and what do they do?
Alpha 2 (also Beta receptors); decrease motility
Which sympathetic receptors are present in sphincter smooth muscle and what do they do?
Alpha 1(also Beta); contraction
Which sympathetic receptors influence gastric secretion and what do they do?
Alpha 2 (also Beta); inhibit secretions
Which sympathetic receptors are present in bladder wall (Detrusser) smooth muscle and what do they do?
Beta 2; relax the smooth muscle
Which sympathetic receptors are present in bladder sphincter (Trigone) smooth muscle and what do they do?
Alpha 1; contract
Where is the exception to the rule that parasympathetic nerves do not innervate vascular beds?
The corpra and small blood vessels of the penis
What disorder are viagra-like drugs used to treat in some women between the 3rd-4th decade of life?
Pulmonary Hypertension
In erection what does Point and Shoot mean?
Parasympathetic/Sympathetic
Which sympathetic receptors are present in salivary glands and what do they do?
Alpha 1; produce viscous secretions
Which sympathetic receptors are present in lacrimal glands and what do they do?
Alpha: secretion
Which parasympathetic receptors are present in lacrimal glands and what do they do?
Muscarinic; copious secretions
When it comes to Eccrine (generalized) sweating what is the receptor used by the sympathetic nervous system and what is causing the sympathetic stimulation?
Muscarinic; Thermoregulation
When it comes to Apocrine (localized) sweating (e.g. head and palms) what is the receptor used by the sympathetic nervous system and what is causing the sympathetic stimulation?
Alpha receptors; mental/psychological stress
Contraction of which muscle is responsible for pupil constriction and what are the receptors that cause this contraction? Which branch of the autonomic nervous system does this.
The Circular Muscle of the iris and Muscarinic receptors are responsible for this response; Parasympathetic branch
Contraction of which muscle is responsible for pupil dilation and what are the receptors that cause this contraction? Which branch of the nervous system does this?
The Radial Muscle of the iris and Alpha 1 receptors are responsible for this; Sympathetic branch
Which muscle of the eye is responsible for movement of fluid out of the globe and lens accommodation for near vision? Which receptors and branch of the nervous system are responsible for this?
The Ciliary muscles, muscarinic receptors and the parasympathetic nervous system
What ion is responsible for NT release from presynaptic terminals?
Ca++
At low therapeuptic doses which branches of the circulatory system does Nitroglycerine preferentially dilate?
Preferentially dilates the veins because there is more NO release and production that occurs in response to Nitro in the veins than in the arteries
What medication is commonly give with Nitroglycerin and why?
Beta blockers; Nitroglycerine will cause the veins to dilate and contribute a decrease in blood pressure. This is turn will cause an increased sympathetic response as the heart tries to maintain BP constant by increasing heart rate (undesired effect). Beta blockers are given to stop this unwanted increase in HR
What effect does intravenous acetylcholine have on blood pressure and why?
It decreases BP because it is a vasodilator and acts on muscarinic receptors on the endothelial cells of vascular beds to produce NO.
What purpose do the muscarinic receptors on the endothelial cells of vascular beds serve and are they activated endogenously or pharmacologically?
Their purpose is unknown since there seems to be no parasympathetic innervation to them. Thus, their main method of activation is pharmacological because the parasympathetic system seems to have little affect.
What two hemodynamic components are related to BP?
Peripheral resistance and Cardiac output
Increases in peripheral resistance in the cardiovascular system are mediated by which sympathetic receptors?
Alpha 1 and Alpha 2
Pulse pressure is related to cardiac output, which is related to contractility of the myocardium, which is controlled by what sympathetic receptor?
Beta 1
What is the major mechanism of attenuated response after stimulation of a postsynaptic terminal with a NT?
Reuptake > Diffusion > Degredation
Which receptors is low dose epinephrine more selective for Alpha or Beta, and which subtype? What does this mean for vasculature the effect on BP?
Beta 2 receptors. This will cause vasodilation and a decrease in BP
Where does epinephrine get released from?
The adrenal medulla
Why do you get a fall in BP at the end of a High Dose Epinephrine response?
Because it starts to act like Low Dose Epinephrine
Which adrenergic receptors dose epinephrine activate?
All the adrenergic receptors: all the Alphas and all the Betas, including Beta 3
What effect does a blocking dose of atropine have on BP?
It has no effect because atropine blocks the effects of the parasympathetic system, but it is the sympathetic nervous system that is responsible for the changes in peripheral resistance and cardiac output that affect BP
What receptors does Isoproterenol work on most?
Beta 2; produces dramatic drops in BP
Beta 1; increases the contractility of myocardium
Does Isoproterenol look like High Dose Epinephrine?
No, it is just the opposite
Compare the receptor profiles for Isoproterenol and Epnephrine.
Isoproterenol affects the same receptors as epinephrine except it does not do the Alpha 1s and Alpha 2s
Explain what mechanism does the chemical Tyramine uses to elicit it very prolonged response?
Tyramine must get into the nerve terminals and gets taken up by reuptake mechanisms and kicks out norepinephrine. This gives a prolonged pressor response
What medication uses a mechanism of action similar to the chemical Tyramine?
Ephedrine; but ephedrine also directly stimulates at nerve terminals in addition to having similar indirect effects that Tyramine does
When do you see hypertension and tachycardia at the same time in a person?
When they are taking Cocaine or amphetamines. Cocaine stimulates sympathetic outflow and has an effect on blocking reuptake of catecholamines
Will Tyramine's effect on BP be changed after pre-treatment with cocaine?
Yes- it will not have it's same effect because it cannot get into the nerve terminal through its reuptake mechanism
What experimental question can Tyramine be used to answer?
Is there a block in reuptake.
What type of receptors does Phentolamine block? Is it competitive or non-competitive?
Alpha 1 and Alpha 2; competitive
Which receptors help provide negative feedback for the release of Norepinephrine? What Alpha blocker stops this appropriate negative feedback and can cause reflex tachycardia?
Pre-synaptic Alpha 2 receptors (in the NTS in particular); Phentolamine because it blocks both Alpha 1 and Alpha 2
What drugs can cause Epinephrine reversal and why?
Alpha blockers, because you have the unopposed vasodilatory and ionotropic responses of Beta 2 and Beta 1 receptors respectively, now that the Alpha receptors in the vascular periphery are blocked
Which receptors does propranolol block? Why would it give you a drop in blood pressure?
Beta 1 and Beta 2 receptors; non-selective beta blocker; Beta 1 controls the increased ionotropic effect seen in heart muscle
What are the two type of Cholinesterase inhibitors?
Carbamates (reversible inhibitor) and Organophosphates (irreversible inhibitor)
What is Physostigmine used to treat and what special boundary does it cross?
It is used to treat cholinergic syndrome and it is able to cross the BBB
What is Neostigimine used to treat and what special barrier can't it cross?
It is used to treat Mysthinia Gravis and it cannot cross the BBB
What drug is only short acting and used to treat MG?
Edrophonium
Which organophosphate is used to treat glaucoma as an anticholinesterase?
Echothiophate
List the organophosphate anticholinesterase war gases in order of faset to slowest enzyme-binding potential.
Soman > Sarin > VX
What does the mneumonic SLUDE or DUMBELSS describe what are they refering to?
They describe the effects of Anticholinesterase poisoning:

Diarrhea
Urination
Miosis/Muscle weakening
Bronchoconstriction
Bradycardia
Emesis
Lacrimation
Salivation
Sweating
In anticholinesterase poisoning, symptoms due to which receptors are the most deadly and why?
The Muscarinic symptoms; they are the most deadly because of respiratory compromise due to bronchoconstriction, bronchial secretions and CNS-respiratory failure
What is the Rx for AChEI poisoning with a Carbamate and how does it work?
Atropine, which also has a wide therapeutic index (compared with that of 2-PAM for Organophosphates), is an antimuscaric agent, so it blocks the receptors
What is the Rx for AChEI poisoning with Organophosphates and how does it work?
2-PAM (Pralidoxime), which has a very narrow therapeutic index (because it has NMJ blocking side effects), but which actually prevents the irreversible binding of organophosphates with AChE (but must be given BEFORE AGING occurs)
Name the drugs that are anti muscarinic.
Atropine, Scopalamine, Ipratropium Bromide, Glycopyrrolate
Which Muscarinic receptors does Atropine work on and what function does it serve in eye exams?
m1-m5; it functions to dilate the pupil by inhibiting parasympathetic tone of the ciliary muscle (Mydirasis and cycloplegia)
What are the natural sources that contain Atropine and Scopalamine?
Atropa belladonna (deadly nightshade) and Jimson Weed
When is atropine contraindicated?
Narrow angle glaucoma
Why are atropines sometimes used before surgery?
Because the salivary glands and other secretory sites are particularly sensitive to muscarinic blockade and they can prevent secretory complications with airways
Compare the CNS effects of Atropine and Scopalamine and why?
Scopalamine has greater CNS effects because it can cross the BBB and Atropine cannot
What are the therapeutic uses of Scopalamine?
Given prophylactically for motion sickness, and for treatment of side effects of Parkinson's Rx's and some antipsychotics
What condition are antimuscarinics use for with regard to heart rate?
Bradycardia; reduces vagal tone and brings heart rate up to a physiological range and keeps more stable
What are some of the effects of anticholinergics on the respiratory system?
Decreased secretion of bronchal mucus, reversal of any vagal-mediated bronchoconstriction
What anticholinergic is given to treat asthma? Why are there no CNS effects?
Ipratropium; it quaternary and gets stuck in the lung, so it cannot get to the brain
What are the 5 cardinal signs of Anticholinergic toxicity?
1) Blind as a Bat
2) Dry as a Bone
3) Red as a Beet
4) Hot as Hell
5) Mad as a Hatter
Provide a summary of the Therapeutic indications for Antimuscarinics.
-Presurgical adjunct to decrease bronchal secretions
-Eye exams
-ICU Tx of sinus bradycardia and AVN block
-Prevent motion sickness
-Asthma
-Parkinsonism
-Antidote to Anticholinesterase
Tubocurarine is the prototypic non-depolarizing NMJ blocking agent. What are the three mechanisms by which it works?
Competitive inhibition at low doses, higher doses can also inhibit ACh release from presynaptic terminals, and very high doses can also block the nicotinic channel
Tubocurarine is often given pre-surgically to facilitate intubation and help immobilize the patient. How do you reverse these effects after surgery is over?
NMJ blockade can be over come with ACh and an AChEI (e.g. neostigmine). Atropine or Glycopyrrolate are also given to prevent muscarinic effects of neostigmine.
Is Tubocurarine specific or selective for NMJs?
Selective! NOT specific..
What is the main depolarizing NMJ blockin agent? Is it an agonist or antagonist?
SuccinylCoA; it is an agonist with rapid onset and rapid hydrolysis (short action).
Describe the three phases of SuccinylCoA action.
Initially- there is depolarization of the muscle end plate and fasciculations can be seen on the skin

PHASE I- continuous depolarization prevents the generation of additional muscle contractions and leads to flaccid paralysis

PHASE II- with continuous administration the end plate hyperpolarizes, but is desensitized to the effects of ACh
What are some of the more prominent adverse side effects seen wtih SuccinylCoA administration?
Hyperkalemia, muscle pain, malignant hyperthermia (occurs with a genetic mutation)
How does Botulinum toxin work?
It degrades SNARE vesicle docking proteins in presynaptic terminals and prevents vesicular release of ACh
What are some therapeutic indications for Botox that are on the rise?
Facial spasms, ocular strabismus, muscle spasticity, muscle pain disorders (OL), headaches (OL), Excessive sweating, BPH (OL)
Which drugs are selective Ganglionic Nicotinic receptor inhibitors? What responses do they elicit and when are they used?
Mecamylamine and Trimethaphan are selective for both Symp. and Paras. ganglia. They block all reflex regulation of heart and BP and produce a complete loss of predominant tone. They are used during a hypertensive crisis or an aortic aneurism.
Does Norepinephrine have greater Alpha effects or greater Beta effects? Which receptor subtype does it have the least affinity for?
Alpha effects; Beta 2
What two sympathetic receptors work together to raise blood pressure and how do they do it?
Alpha1 (increased peripheral resistance) and Beta 1 (increased chronotropic/ionotropic efffect)
When sympathetic tone on the heart is induced what is the reflex pathway that normally occurs and which adrenergic receptors are involved where?
Alpha‐1 stimulation results in an
extreme elevation of peripheral resistance thus increasing blood pressure. This markedly increased
blood pressure leads to baroreceptor feedback that inhibits the beta‐1 stimulation. Thus what the patient is left with is an increased blood pressure as a result of increased peripheral vascular
resistance.
When blood pressure is high because there is increased sympathetic tone leading to increased vascular resistance, is there high or low perfusion to the organs?
There is low perfusion, even though BP numbers may appear normal, there is less blood flowing to where it needs to
Epinephrine is a non-selective adrenergic agonist, but at low doses which type of receptors, Alpha or Beta, does it have the greatest affinity for?
Beta
With larger doses of Epinephrine which receptors, Alpha or Beta, are seen to have greater activity?
Alpha. Thus, with higher doses of epinephrine you can induce norepinephrine-like symptoms.
What are the most common indications for epinephrine?
Cardiac arrest and Anaphylaxis
Are dopamine's effects on the body constant or do they change with the dose given?
Dopamine's effects are dose dependent
Which receptors are preferentially stimulated and what response is seen when Dopmine is given in a low dose?
Dopamine receptors; Dopaminergic effect (improving renal and mesenteric function)
Which receptors are preferentially stimulated and what response is seen when Dopmine is given in a medium range dose?
Beta receptors; Beta effects (chronotropic and inotropic effects on the
heart)
Which receptors are preferentially stimulated and what response is seen when Dopmine is given in a large dose?
Alpha receptors; Alpha effects (aka, norepinephrine effects on vasculature, “leave em
dead.”
When is Dopamine indicated?
In cardiogenic or septic shock
Which type of adrenergic receptors does Isoproterenol work on?
Beta; it is a non-selective beta agonist and has no effect on alpha receptors
What kind of effect does Isoproterenol have on BP?
It decreases Diastolic BP due to Beta 2 effects.
When is Isoproterenol indicated?
It is useful in the management of bradycardia, non severe
heart block or cardiogenic shock, until a more definitive treatment can be initiated (such as
transcutaneous pacing)
How is Isoproterenol degraded?
Isoproterenol is degraded by COMT
What intracellular enzyme exists in the presynaptic terminals associated with mitochondria degrades norepinephrine?
Monoamine oxidase (MAO); Catecholamine O-methyl Transferase (COMT)
How much higher is [Ca++] in the extracellular fluid than in the cell?
1000X higher extracellular
What organ and tissue is parasympathetic innervation particularly important for? What molecular intermediate does it work through?
It is important in the corpra of the penis for production of an erection. It works through increasing the production of NOS and NO, which vasodilates and allows more blood flow
What are the mechanisms of action of Cocaine?
Blocks the reuptake of norepinephrinefrom synaptic cleft, and impairs conduction of SA rhythm in the heart...increased action of norepi and impaired cardiac rhythm can lead to VFib in some people
How do direct acting agonists of the sympathetic system work? What are they?
They directly stimulate adrenergic receptors; they are Norepinephrine, Epinenphrine, and Phenylephrine
How do indirect acting agonists of the sympathetic system work? What are they?
They stimulate norepinephrine to be released from axon terminals and work on adrenergic receptors; they are Tyramine, Ephedrine, (dex)Amphetamine
What is the rate limiting step in the biosynthesis of Norepinephrine and Epinephrine? Molecule negatively regulates this step?
The cytosolic conversion of Tyrosine to Dopa via tyrosnine hydoxylase; Norepinephrine negatively regulates this step.
What is the name of the condition associated with a secondary form of hypertension that is caused by a catecholamine secreting tumor originating most commonly in the spinal column? How do we limit catecholamine synthesis in these patients?
Pheochromocytoma; we treat this by inhibiting the rate limiting step of Tyrosine conversion to Dopa and the progressive synthesis of catecholamines.
Dopamine is converted to Norepinephrine what compartment of the neuron? how does it get there? and by what enzyme is it converted?
In the NT vesicle; got there by VMAT in exchange for H+ ions; by dopamine beta-hydroxylase
What is VMAT, its action, and which drug blocks its action?
VMAT is the Vesicle MonoAmine Transporter, which imports dopamine into presynaptic vesicles in exchange for H+ ions for conversion into norepinephrine; Recerptine blocks the action of this transporter
What is the main adverse side effect of Reserprine and why does it cause it?
The main adverse side effect oof Reserprine is depression; Resprine causes depression because it inhibits VMAT in the CNS neurons and subsequent dopamine ---> norepinephrine conversion just like it does in the sympathetic neurons leading to the alpha receptors of the vascular smooth muscle
MAO and COMT are both catecholamine degrading enzymes. Where is each found with respect to intra/extracellular compartments?
MAO is found intracellularly, bound to the mitochondrial membrane. COMT is found in the extracellular space in the synaptic cleft
What are two enzymes found in the terminals of sympathetic nerves are targeted to knock down the production of norepinephrine?
Tyrosine hydroxlase (Tyrosine --> Dopa) and VMAT (Vesicle MonoAmine Transporter)
If Alpha receptors are close to the nerve terminal which subtype are they? If they are far away which subtype are they?
If they are close to the nerve terminal they are Alpha 1, if they are far away they are Alpha 2
How do Alpha receptors exert their action in smooth muscle cells?
They increase the cytoplasmic levels of Ca++ by releasing it from intracellular stores and increasing permeability from the extracellular compartment
Why are Calcium channel blocking drugs such important antihypertensive drugs?
Because they affect the sympathetic tone on blood vessels so effectively (and they affect ionotropic effects in the heart?)
What four classes of drugs make up ~90% of the prescribed medications for patients that suffer from hypertension? What four mechanisms to they promote?
Beta blockers (Decreased cardiac output), Ca++ Channel blockers (decreased sympathetic vasomotor tone), ACE inhibitors (decrease vasconstriction with ANGII, the most powerful constrictor known), Thiazide diuretics (decrease blood volume)
Which enzyme does Cocaine Block at nerve terminals?
NET- norepinephrine enzyme transporter; this block reuptake of norepi
How do the indirect acting agents, Ephedrine and Amphetamine work at the nerve terminal?
NET(Norepinephrine Enzyme Transporter) takes up the indirect acting agent and Norepinephrine is displaced from the cytosol in to the synaptic cleft where is can elicit its response
How does Cocaine effect the action of indirect acting agents on the sympathetic system? (increase, decrease, no change?)
Cocaine DECREASES the action of indirect acting agents because cocaine targets NET and this is the same transporter that takes up the indirect acting agents
What purpose does the autoreceptor on the nerve terminal of sympathetic nerves serve, what type of receptor is it and what NT does it bind?
The purpose is to attenuate the release of Norepinephrine; the receptor is and Alpha 2 receptor
How can blocking some subtypes of Alpha receptors lead to an exaggerated response of Norepinephrine release?
This is because there are Alpha 2 receptors on the presynaptic terminals that are involved in a negative feedback loop, that decrease the release of norepinephrine. When they are blocked, so is this feedback
By definition, which receptors are located at the posynaptic end of the nerve terminal close to where the sympathetic nerves terminate 1) at vascular smooth muscle and 2) at the heart
by definition 1) Alpha 1 and 2) Beta 2
How does feedback on the presynaptic Alpha 2 autoreceptor decrease the release of Norepinephrine?
By decreasing the ability of Ca++ to influx across the cell membrane during stimulation and thus inhibiting the exocytotic process and vesicle release of NE
What is the mechanism of action that Calcium elicits to increase smooth muscle contraction?
Ca++ bind with Calmodulin and phosphoylates myosin light chains to produce smooth muscle contraction
How do Alpha receptors lead to the increase of Ca++ intracellularly?
Stimulation of Phosphlipase C ---> IP3 ----> releases intracellular Ca++ stores and activates PKC at the membrane along with Ca++
When we are stimulating Alpha 2 presynaptic receptors pharmacologically to suppress sympathetic tone, where is the anatomically site of action?
The Nucleus Tractus Solitarius in the brain stem
What change in the Frank-Starling curve indicates a positive ionotropic response?
A shift in the curve up and left indicates a positive ionotropic response
What change in the Frank-Starling curve indicates a negative ionotropic response?
A shift in the curve down and to the right indicates a negative ionotrpoic response
What causes positive ionotropic effects in the heart?
Sympathetic stimulation, Beta 1 agonists, Digoxin
What causes negative ionotropic effects in the heart?
Heart Failure; or excess Beta Blockers
How do Beta 2 receptors stimulate vasodilation?
They stimulate the production of cAMP which acts to phosphorylate proteins in the membrane of the sarcoplamic reticulum. These phosyphorylated proteins take up the calcium from the cytosol and lead to decreased contraction and increased dilation
What are the effects seen from stimulation of A1 postsynaptic adrenergic receptors?
Vasoconstriction, mydriasis, contractions of GI and bladder SPHINCTERS
What are the effects seen from stimulation of A2 postsynaptic adrenergic receptors?
Vasoconstiriction (blood vessels)
What are the effects seen from stimulation of A2 presynaptic adrenergic receptors?
Decreases central sympathetic activity and inhibits the release of norepinephrine from sympathetic nerves (NTS & nerve terminals)
What are the effects seen from stimulation of B1 postsynaptic adrenergic receptors?
(+) Ionotropic and (+) chronotropic effects in the heart; there are also arrhythmogenic effects that can be observed especially when stimulated with high doses of agonist (these can be good if there is a heart block and no conduction, but usually bad and produce ectopic conduction)
What are the effects seen from stimulation of B2 postsynaptic adrenergic receptors?
Vasodilation, bronchodilation, mobilization of glucose and possibly fat stores (mechanisms mediated through cAMP)
What are the effects seen from stimulation of B3 adrenergic receptors?
Mobilization of fatty acids and breakdown of fat cells and adipose tissue
What is the effect produced by Phenylephrine, which receptors does it stimulate and when is it indicated?
Phenylephrine produces a long acting and long lasting rise in BP due to action on A1 receptors; this is indicated during spinal and general anesthesia, vascular shock, drug induced hypotension, overcoming paroxysmal supraventricular tachycardia
Which receptors does Norepinephrine (Levophed) work on and why can't it be used in an acute asthmatic attack?
It stimulates A1, A2, B1 >> B2; its effects on B2 are so minimal that it is not effective in an acute asthmatic attack
Why do you get a smaller increase in BP when you use Epinephrine vs. Norepinephrine?
This is because Epinephrine simulates B2 very well in addition to stimulating A1, A2, and B1 like NE. The vascular beds in Skeletal muscles are big and laden with B2 receptors, so when they dilate they can have a profound effect on BP. This attenuates the response of BP to Epi compared to the response of BP to norepi.
Explain what type of shock is Isoproterenol good for treating and which type is it not? Why and how does it have to do with the receptors it stimulates?
Isoproterenol stimulates B1 and B2 receptors only. Thus it is, Good for Cardiogenic shock where there is low cardiac output and increased output is needed. It is bad for anaphylactic shock or Gram (-) septic shock that are more due to vasodilation. Because of its powerful effects on the B2 receptors in the vascular beds of skeletal muscle it could exacerbate the problem
What is stimulation of D1 dopamine receptors used for?
To produce vasodilation of mesenteric and renal vessels to promote prefusion of mesentery and kidneys;
What two types of receptors does Dopamine affect at higher concentrations?
B1s and A2s, thus it helps support BP during shock and is used in acute heart failure
What adrenergic agent stimulates only B1 receptors and is used in cardiogenic shock and heart failure?
Dobutamine
What is the major form of metabolism of Norepinephrine from the synaptic cleft?!?
REUPTAKE!!!!
What is the major metabolite of Norepinephrine and epinephrine that is produced in the liver (via MAO) from excess metanephrine and normetanephrine (metabolites that when epi and norepi are increased are found in the urine)? What condition would you expect to find in this patient?
VMA; tremendously increased in pheochromocytoma tumor
Where do we find Catechol-O-methyltransferase (COMT) in cells?
Extracellularly, associated with membranes
What intracellular enzyme degrades Tyramine?
MAO
What happens to the response elicited by Tyramine when Phenelzine (an MAO inhibitor) is given?
It goes way up! Because MAO degrades neither Tyramine nor NE now!
What foods contain Tyramine?
Red wine, aged cheeses, smoked oysters
What are MAO inhibitors traditionally used for?
Depression
What do patients taking MAO inhibitors have to be careful of concerning diet?
They must not consume foods with large amounts of Tyramine in them! (or take ephedrine)
What is the mechanism by which MAO inhibitors can help lower BP?
By producing the false NT Octopamine. When low amounts of Tyramine in the diet are taken up by VMAT on the membranes of vesicles in nerve terminals, this Tyramine gets converted to Octopamine (an NT much weaker than NE, and an A2 blocker) by DBH. Octopamine helps lower blood pressure
Do MAOIs have a bigger effect on NE or indirect acting adrenergic agents?
Indirect acting!; no change is observed with the direct acting agents
How much of the total NE pool in the vesicles of nerve terminals is released as a result of indrect sympathetic agonist action?
about 3%
Amphetamine, Ephedrine, Tyramine:
Of these drugs which are direct acting, indirect or both? If there is a direct effect, which adrenergic receptors are stimulated?
Amphetamine- both indirect and direct action; simulates Alpha receptors
Ephedrine- both indirect and direct action; stimulates both Alpha and Beta receptors
Tyramine- indirect action only
What is Tachyphylaxis?
Tachyphylaxis refers to decreased response to the action of indirect acting adrenergic agonist upon successive dosing due to exhaustion of cytosolic NE
What is the other drug that begins with an "I" that works on sympathetic nerve terminals through the same mechanism of action as Cocaine?
Imipramine
What effect does Reserpine pretreatment have on the action of indirect acting adrenergic agonists? WHY?
It blocks the effects of indiirect acting adrenergic agonists because those drugs work through kicking out NE that is stored in the presynaptic vesicles. Reserpine blocks VMAT, the transporter that brings Dopamine into those vesicles, in order to make NE
How does Bertylium exert its action at the end of sympathetic nerve terminals?
It uncouples the release of vesicular NE from the action potential (Ca++ mediated event). Also used as a K+ channel blocker.
What four other drugs does Guanethidine share mechanisms of action with?
Tyramine (kicks out NE after being taken up by NET)

Cocaine (blocks NET reuptake of NE)

Reserpine (blocks VMAT and vesicular NE production)

Bertylium (dissociates vesicular release from action potential)
What are the effects of Guanethidine on BP?
Initially it acts like Tyramine and Cocaine and raises BP due to vasocontriction.

Later it acts like Reserpine and Bertylium and lowers BP due to vasodilation.
Why can't we give ACE inhibitors to women?
Because ANGII is a growth factor, so anything that would inhibit its effects iis absolutely contraindicated in pregnant women
What is a substitute medication that we give to pregnant women in place of an ACE inhibitor? How does it work?
Alpha-methyldopa; it works much like the MAOI + Tyramine mechanism ---> it produces alpha-methylnorepinephrine in vesicles, which acts as a false NT and stimulator of presynaptic A2 receptors at the nerve terminals and NTS (negative feedback on NE release)
Which receptors do each of these drugs work on?
1) Dobutamine
2) Albuterol
3) Terbutyline
4) Phenylephrine
5) Clonidine
6) Dopamine
1) B1
2) B2
3) B2
4) A1
5) A2
6) D1>>B1 and A2
What do you give a patient with Gram (-) sepsis?
Dopamine; but if you have to continually increase the concentration to try and produce effects there is risk of extreme vasoconstriction and arrhythmias
What percentage of the heart's fuel comes from free fatty acids?
~70%
What drug is the most potent effector of B1 and B2 receptors because of it's two methyl groups the end of the molecule?
Isoproterenol
What is tocolytic action (B2 manipulation)?
Tocolytic action means relaxation of the Urterus- can stop contraction if necessary, such as in the case of early labor
What type of signal transducing proteins do Beta and Alpha receptrors work through? Which subtype specifically for Betas regardless of whether they contract or relax? Which for postsynaptic A2s?
They both work through G protein signaling mechanisms. Betas work through Gs proteins and postsynaptic A2s work through Gi proteins (but, still have net increase on cytoplasmic Ca++)
How do B2 receptors work to produce a smooth muscle relaxation? What is the key ion in the cell and does its cytoplasmic level increase or decrease?
B2 works by decreasing the cytosolic levels of Ca++, and facilitating its uptake into intracellular stores. Increased levels of cAMP activate cAMP dependent kinase phosphorylates special proteins on the SR and Ca++ is taken up into store
How do B1 receptors work to produce increased contractility? What is the key ion in the cell and does its cytoplasmic level increase or decrease?
B1 works by increasing the cytosolic levels of Ca++, and facilitating its release from intracellular stores. Increased levels of cAMP activate cAMP dependent kinase phosphorylates special proteins on the SR and Ca++ is released from store. There is also more Ca++ that enters from the extracellular compartment through the L-type channel
How do postsynaptic A2 receptors work to produce smooth muscle contraction? What is the key ion in the cell and does its cytoplasmic level increase or decrease?
Postsynaptic A2 receptors stimulat Gi protein activity and decrease levels of intracellular cAMP. Still, there ends up being net increase in cytosolic levels of Ca++ and thus increased contraction of smooth muscle.
Which medications are non-selective Alpha receptor agonists only?
None
Which medications are non-selective Beta receptor agonists only?
Isoproterenol
Which medications are non-selective Beta and Alpha receptor agonists only?
Epinephrine
Which medications are non-selective Alpha receptor agonists and Selective Beta receptor agonists only?
Norepinephrine; only B1
Which medications are selective Beta1 receptor agonists only?
Dobutamin
Which medications are selective Beta 2 receptor agonists only?
Tertbutyline and Albuterol
Which medications are selective Alpha 1 receptor agonists only?
Phenylephrine and Methoxamine (rebound vasodilation can be a problem)
Which medications are selective Alpha 2 receptor agonists only?
Clonidine and Methyldopa (can use in preeclampsia- Hypertension during preganancy)
What alpha 1 stimulator is commonly used as a nasal decongestant?
Phenylephrine
Which adrenergic stimulators are considered Catechols?
Catechol, Phenylethylamine, Norepinephrine, Epinephrine, Isoproterenol, Dopamine
Which adrenergic stimulators are not considered Catechols?
Phenylephrine, Methoxamine, Ephedrine, Amphetamine
What drug would you use to test whether or not the effect of an adrenergic stimulator was direct or indirect?
Cocaine, because it definitively only blocker the action of an indirect stimulator
What are the major uses for Beta Blocking drugs?
Hypertension, Ischemic Heart Disease/Angina, CHF,
Which drugs are Non-selective Alpha blocking agents?
Phentolamine and Phenoxybenzamine
Which drug is a Non-selective Beta blocking agent?
Propranolol
Which drug is a selective A1 blocking agent?
Prazosin
Which drug is a selective B1 blocking agent? What is it used for?
Metoprolol; antianginal, antihypertensive, asthmatic patients with caution, Hypertensive with CHF, CHF
Which drug is a selective A2 blocking agent?
Yohimbine
Which drug is a selective B2 blocking agent?
Butoxamine
Do the Non-specific Alpha blocking agents have much use in medicine today? Why or why not? Which is competitive and which is non-competitive? What special case could they be used for?
They do not have much use today, mostly because they have not proven to be that effective at reducing hypertension and they have undesirable side effects (Reflex tachycardia). Phenoxybenzamine (long-acting)is a non-competitive inhibitor and Phentolamine (short-acting) is a competitive inhibitor. They could be used in the special case of Pheochromocytoma.
What are two main uses that A1 blockers are used for? What is the main example of this class?
Hypertension and Benign Prostatic Hyperplasia (BPH); the main example is Prazosin
What purpose do A2 receptor blockers serve?
They block the A2 receptors in the NTS so they there is less feedback inhibition of Norepinephrine and you get a better reflex response when the sympathetic system is stimulated say in the case of blood flow and a change in posture
What are the three drugs we can use to treat Glaucoma and which classes do they belong to?
Pilocarpine (Cholinomimetic), Echothiophate (AchEI), Propranolol (Beta-blockers)
Why do Non-selective Alpha blockers give reflex tachycardia and the Selective A1 blockers do not?
Because with the Non-selective Alpha blockers the negative feedback on Norepinephrine of the presynaptic A2 receptors in the Sinus Node is not in tact, whereas with the selective A1 blockers it is.
What is Dyslipidemia?
This is a general term associated with high cholesterol or high triglyceride levels in plasma
What are the two major sequelae of hyperlipidemia?
Acute pancreatitis and atherosclerosis
Explain what each of the three carrier molecules are used for: Chylomicrons, vLDL, LDL.
Chylomicrons are formed in the intestines and are for transporting TGs of dietary origin to the liver. vLDL is secreted by the liver and provides a means for TG from the liver to be exported to the tissues. LDL transports cholesterol from the liver to the blood stream, and high levels of this are associated with increased risk of atherosclerosis and Coronary Artery Disease
Where does HDL transport cholesterol to and from?
HDL transports cholesterol from the periphery back to the liver
Describe Familial Hypercholesterolemia and what its underlying mechanism is. How would you treat it?
Familial Hypercholesterolemia is an autosomal dominant trait which produces a defect in the affinity of LDL receptor. Homozygous individuals can have serum cholesterol levels as high as 1000mg/dl (This would lead to childhood development of coronary disease). Niacin and Atorvastatin are beneficial, but binding resins have little to no effect depending on LDL receptor activity.
Describe Familial Ligand-Defective Apolipoprotein B Dyslipidemia.
This comes from a defect in ligand region of apo B100 region of LDL; impairs the endocytosis of LDL leading to moderately severe hypercholesterolemias
How do we treat Familial Ligand-Defective Apolipoprotein B Dyslipidemia.
Statins (upregulators of LDL receptors) have variable effects; Niacin produces beneficial effects by reducing vLDL plasma levels
What enzyme determines the rate of Triglyceride clearance?
Lipoprotein lipase.
What 4 conditions are associated with secondary dyslipidemias?
Diabetes(both high TG and cholesterol), Hypothyroidism(high cholesterol, but normal TG), BC Pills(high cholesterol and TG), Drug Induced Dyslipidemia
How does Niacin work to improve a person's lipid profile?
It decreases LDL and vLDL levels, and raises HDL levels; the mechanism involves the inhibition of vLDL secretion there by reducing LDL levels. Also lowers both TG and Lp(a) levels
What are some of the side effects of Niacin? Drug interactions?
Niacin flushing, hyperuricemia, hepatic dysfunction; works synergistically with ganglionic blockers leading to orthostatic hypotension
Name two medications that are Fibric Acid derivatives?
Gemfibrozil and Fenofibrate
What is the mechanism of action of Gemfibrozil and Fenofibrate? What conditions are they used to treat?
They reduce vLDL levels and increase the activity of lipoprotein lipase, and also lower TGs and raise HDL. These medications are used to treat Hypertriglyceridemias in which vLDLs predominate and dysbetalipoproteinemia
What are some of the side effects of Fibric Acid derivatives?
Myopathy, arrhythmias, elevation of liver enzymes, cholesterol gallstones, increased anticoagulant effect of coumarin and indandeiones.
What medications are members of the class Bile Acid Binding Resins?
Colestipol and Cholestyramine
How do Colestipol and Cholestyramine work?
These medications bind bile acids in the intestine forming a complex that is excreted in the feces. This prevents the reabsorbtion and recycling of bile acids and so the body increases the oxidation of cholesterol to bile acids to replenish what was lost. This decrease in cholesterol results in an increase in the number of LDL receptors, which thereby decrease the serum LDL levels
What are some of the side effects of the bile acid binding resins?
constipation, may interfere with normal fat digestion and absorption, may prevent the absorption of fat soluble vitamins such as A, D, E, and K
What do statins do? How do they work?
Statins are structural analogs of HMG-CoA and are competitive inhibitors of HMG-CoA reductase; this is the enzyme that mediates the first committed step in sterol biosynthesis; This leads to an induction of High Affinity LDL receptors, thus a lowering of serum LDL levels (may also have an effect on C-reactive protein)
Why are Statin drugs usually taken at night? Which Statin is an exception and why?
This is because cholesterol metabolism usually occurs, and is at its highest, at night time. The drug is not taken to maintain constant levels because tolerance is a concern. The exception to this is the drug Atorvastatin. This can be taken at other times because it has such a long half life that plenty of drug is left around at night
Which Statins are special in that the are pro-drugs and must be activated in the intestine vs. the rest of the Statins, which are ingested in active forms?
Lovastatin (Mevacor) and Simvastatin (Zocor) are inactive prodrugs that are activated in the GI tract
Which Statin is nearly completely absorbed?
Fluvastatin (Lescol)
What metabolic liver effect is observed to be high in Statins?
There is a high first pass liver effect
What is the average half life associated with most Statin drugs? Which is the exception and what is its half life?
The average half life of a Statin is 1 to 3 hours; the exception to this is Atorvastatin (Lipitor) which has a half life of about 14 hours
What are some common side effects of taking Statins? What is a less common effect that is more serious?
It is common to see 3x elevation of liver enzymes in the serum, which does not need to alarm for liver toxicity unless there is already underlying liver disease in the patient. A less common side effect is RHABDOMYOLYSIS which may occur leading to myoglobinemia and subsequent RENAL FAILURE
Drugs interaction that effect what molecules in the liver must be considered when prescribing Statins?
Those drugs that affect Cytochrome metabolism; drugs that inhibit or compete for CYP3A4 or CYP2C9 will increase Statin levels in plasma; drugs that induce CYP450s will reduces Stain levels in plasma
Can we give Statins to pregnant women?
NO! they have TERATOGENIC potential!!!
How does Ezetimibe work? Is it a prodrug or already active?
This is a prodrug that works by decreasing GI uptake of cholesterol (both dietary and biliary secreted cholesterol). This reduces the tightly regulated cholesterol pool in the liver resulting in invreased synthesis of high affinity LDL receptors and subsequent removal of LDL from the blood. This lowers serum LDL and TGs!!
What is the toxicity associated with Ezetimibe and HMG-CoA reductase inhibitors (Statins)?
They may increase hepatic toxicity when administered together.
Why is it standard therapy to initiate HMG-CoA Reductase Inhibitor Therapy immediately after myocardial infarction, irrespective of lipid levels?
This is because in addition to inhibiting cholesterol synthesis, these drugs decrease oxidative stress, as well as vascular inflammation and promote increased stability of atherosclerotic lesions.
What are the effects of Fibric acid derivatives on the lypolysis of lipoprotein TGs and in adipose tissue? What receptors do they bind to?
Lipoprotein TG lypolysis is increased, while intracellular lipolysis in adipose tissue is decreased. Fibric acid derivatives function as ligands for the nuclear transcription receptor "peroxisome proliferator-activated receptor alpha (PPAR-alpha)"
What are the lipid molecules and receptors that each of these drug classes works to reduce or increase:
1) Bile acid binding resins
2) Niacin (Nicotinc acid)
3) Fibric acid derivatives
4) Statins
1) Isolated decrease in LDL and IDL; increases LDL receptor
2) Decreases in vLDL, LDL, and TGs; increases HDL (most effective agent at doing this)
3) Decreases vLDL, LDL, and TGs; slight HDL increase
4) Decreases vLDL and LDL; increases LDL receptors
Explain Variant Angina? What is another name for this condition?
Variant angina is a coronary insufficiency syndrome due to VASOSPASM (can appear in the absence of CAD). This may be an effect due produced by atherosclerosis on vasomotor tone. Attacks often occur during rest (e.g. at night). It is also called PRINZMETAL'S ANGINA.
What is the pain and ischemia associated with Unstable Angina due to?
Platelet aggreagation at fractured plaques and some vasospasm
What are the main therapeutic features of Nitrates, Calcium Channel blockers and Beta Blockers?
Nitrates are vasodilators with a large preference for veins; Calcium Channel blockers are vasodilatory and cardiac depressants; Beta blockers serve as cardiac and sympathetic tone depressents
Why do organic nitrates (e.g. Nitroglycerin) work through vasodilation preferably of the Veins? What is the theory behind the mechanical heart change of why Nitros work? What is a counter-productive side effect that they can elicit?
Nitroproduces a greater effect on the veins than the arteries because there is more enzyme that converts nitroglycerin to NO in venous smooth muscle vs. atertial. Venodilation results in decreased preload, which leads to decreased work. Decreased work leads to decreased O2 demand less ischemia; Reduction of afterload which can be produced by high doses and arterial dilation can elicit a counter productive reflex tachycardia
Explain the molecular mechanism and pathway that accounts for the action of Organic nitrogen administration.
Nitroglycerine is converted to and stimulates NO production in endothelial cells. NO acts stimulates Guanylyl cyclase which converts GTP to cGMP. cGMP facilitates dephosphorylation of Myosin-LC (no longer interacts well with actin) and protein kinase mediated intracellular Calcium decrease
What are two effects that Nitrates have on the coronary circulation?
1) They redistribute coronary blood flow and can selectively increase blood flow to ischemic areas, with overall blood flow not being increased.
2) There is a reversal of coronary vasospasm
What strategy is employed with patients taking nitrates, to avoid tolerance?
Nightly nitrate free gaps; not taking nitrates at night
Describe some of the side effects and drug interactions seen with Nitrates.
Side effects: Orthostatic hypotension, headache, dizziness, reflex tachycardia, tolerance

Interactions: when taken within 24 hrs of Sildenafil (Viagra) can produce >25mmHg drop in blood pressure
What is a mechanism of action associated with Beta blockers (Propranolol) with regard to oxygen consumption?
They reduce heart rate and force of contraction of the heart, as well as reduce blood pressure during exercise. All this decreases the work of the heart and the O2 demand.
What type of angina are Beta blockers (Propranolol) indicated for? What type are they contraindicated for?
They are used for prophylaxis of exertional angina only (keeps systolic BP and HR low = low O2 demand); They are contraindicated for Variant Angina which is more due to vasospasm (Beta blockers could exacerbated the problem)
Why are Beta blockers frequently administered with Nitrates?
Because a side effect of Beta blockers is increased end-diastolic volume and ejection time (increases O2 demand), Nitrates are often coadministered to decrease this end-diastolic volume and preload
What is the gold standard of treatment for OD of Beta blockers and toxic exposure on heart rate? How does this treatment work?
Glucagon; it works because it has receptors in the heart like Beta receptors that work through a similar pathway and has similar effects as the Beta receptors
Describe the channels that Calcium Channel blockers act on in cardiac and vascular smooth muscle in order to produce their effect of decreased contractilty and vasodilation.
The drugs block calcium influx via L-type channels in cardiac and vascular smooth muscle
Which Calcium Channel blocker is "vascular selective"? What are one of the problems associated with these vascular selective dihydropyridines?
Nifedipine (Procardia); one of the problems with these types of Ca++ blockers is reflex tachycardia
What are the three main Ca++ channel blockers that we learned about? When are they commonly used and when are they contraindicated?
Nifedipine, Diltiazem, Verapamil; they are commonl used as antiarrhythmic agents, they are options to Beta Blockers in asthmatics and useful if patients have A.Fib; they are contraindicated in decreased AV conduction (CHF, AV-block), avoid combining with Beta blocker (decreased HR/decreased SV)
What is the most important physical mechanism by which the Ca++ Channel Antagonists work (affects arteries more than veins)?
Decrease O2 demand through decreased HR, Contract., TPR, BP and increase coronary blood flow (useful in vasospastic angina)
What type of Ca++ blockers are less effective as a monotherapy and why?
The Dihydropyridine type CCBs because they have vascular selective resistance
Explain why the drug combination of Nitrates and Beta blockers is beneficial for patients with Angina?
They work by different mechanisms of action so their effects are additive. Beta blockers prevent reflex tachycardia of nitrates and nitrates reduce increased end-diastolic volume from Beta blockers
Explain what Ranolazine is used for and how it works.
Ranolazine is an alternative antianginal drug for those who can't tolerate other antianginal's side effects. It works by shortening the action potential duration by blocking the surface membrane Na ion channels. This decreases intracellular Ca++ and O2 demand.***Higher doses increase QT by blocking the Potassium Kr channels
What is Clopidogrel and how does it work?
Clopidogrel is a pro-drug whose action may be related to adenosine diphosphate (ADP) receptor on platelet cell membranes. The specific subtype of ADP receptor that clopidogrel irreversibly inhibits is P2Y12 and is important in platelet aggregation and the cross-linking of platelets by fibrin. The blockade of this receptor inhibits platelet aggregation by blocking activation of the glycoprotein IIb/IIIa pathway.
Which two hypolipidemic drug classes are not combined because of exacerbation of potentially harmful side effects?
Statins and Fibrates